Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 288
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 579(7798): 260-264, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32132711

RESUMEN

The production of pore-forming toxins that disrupt the plasma membrane of host cells is a common virulence strategy for bacterial pathogens such as methicillin-resistant Staphylococcus aureus (MRSA)1-3. It is unclear, however, whether host species possess innate immune mechanisms that can neutralize pore-forming toxins during infection. We previously showed that the autophagy protein ATG16L1 is necessary for protection against MRSA strains encoding α-toxin4-a pore-forming toxin that binds the metalloprotease ADAM10 on the surface of a broad range of target cells and tissues2,5,6. Autophagy typically involves the targeting of cytosolic material to the lysosome for degradation. Here we demonstrate that ATG16L1 and other ATG proteins mediate protection against α-toxin through the release of ADAM10 on exosomes-extracellular vesicles of endosomal origin. Bacterial DNA and CpG DNA induce the secretion of ADAM10-bearing exosomes from human cells as well as in mice. Transferred exosomes protect host cells in vitro by serving as scavengers that can bind multiple toxins, and improve the survival of mice infected with MRSA in vivo. These findings indicate that ATG proteins mediate a previously unknown form of defence in response to infection, facilitating the release of exosomes that serve as decoys for bacterially produced toxins.


Asunto(s)
Proteínas Relacionadas con la Autofagia/metabolismo , Toxinas Bacterianas/metabolismo , Exosomas/metabolismo , Células A549 , Proteína ADAM10/metabolismo , Animales , Toxinas Bacterianas/farmacología , Supervivencia Celular/efectos de los fármacos , ADN Bacteriano/farmacología , Exosomas/efectos de los fármacos , Exosomas/ultraestructura , Femenino , Células HEK293 , Humanos , Masculino , Staphylococcus aureus Resistente a Meticilina/patogenicidad , Staphylococcus aureus Resistente a Meticilina/fisiología , Ratones , Ratones Endogámicos C57BL , Infecciones Estafilocócicas/mortalidad
2.
Biochim Biophys Acta Gen Subj ; 1861(1 Pt A): 3365-3377, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27590109

RESUMEN

BACKGROUND: Chemokines play key roles in immune homeostasis and inflammatory response. Considering the role of Ccl20 and Toll-like receptor 9 (TLR9) in gut homeostasis and inflammatory bowel disease (IBD), regulation of Ccl20 by bacterial DNA, the TLR9 ligand, merits in-depth studies. METHODS: We analyzed Ccl20 expression in various epithelial cell (EC) lines by q-PCR and ELISA. In-vivo expression was investigated in isolated murine colonocytes by immunoblotting. Transcriptional regulation of Ccl20 was studied by reporter assays, gene knock-down, electrophoretic mobility shift assay and chromatin immunoprecipitation. Activation of upstream kinases was checked by immunoblotting. RESULTS: We showed low levels of Ccl20 expression in mouse colonic ECs, but marked induction by in vivo treatment with bacterial DNA. This corroborated with persistent Ccl20 induction in different EC lines. We found involvement of MAP-kinases during the early hours after stimulation, and a novel AP-1site (-252bp) regulated the expression in colonic ECs. More importantly, mutually exclusive transcriptional regulation by AP-1 (cjun/cfos) and non-canonical NF-κB (RelB/p52) downstream of MEK-ERK and NIK-IKK-α-NF-κB2 (p100) phosphorylation, respectively was responsible for persistent Ccl20 expression in the colonic cells, while canonical NF-κB isoforms played no role. CONCLUSIONS: Persistent Ccl20 induction by TLR9 in colonic ECs involves early and delayed activation of two independent signaling pathways. This is the first report of non-canonical NF-κB activation and Ccl20 expression in the colonic ECs by TLR9. GENERAL SIGNIFICANCE: Our study will help to better understand immune regulation by Ccl20 in the intestine and may be exploited for future development of novel therapeutics against IBD.


Asunto(s)
Quimiocina CCL20/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , FN-kappa B/metabolismo , Transducción de Señal , Receptor Toll-Like 9/metabolismo , Factor de Transcripción AP-1/metabolismo , Animales , Línea Celular , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Quimiocina CCL20/metabolismo , Quimiotaxis/efectos de los fármacos , ADN Bacteriano/farmacología , Células Dendríticas/citología , Células Dendríticas/efectos de los fármacos , Células Dendríticas/metabolismo , Enterocitos/efectos de los fármacos , Enterocitos/metabolismo , Epitelio/efectos de los fármacos , Epitelio/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/metabolismo , Humanos , Intestino Delgado/efectos de los fármacos , Intestino Delgado/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Ratones , Modelos Biológicos , Factor 88 de Diferenciación Mieloide/metabolismo , Subunidades de Proteína/metabolismo , Transporte de Proteínas/efectos de los fármacos , Factor 6 Asociado a Receptor de TNF/metabolismo , Factores de Tiempo , Transcripción Genética/efectos de los fármacos , Activación Transcripcional/efectos de los fármacos , Activación Transcripcional/genética
3.
Fish Shellfish Immunol ; 62: 349-355, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28119146

RESUMEN

In addition to the crucial roles in coordinating antiviral immune responses, type I interferons (IFNs) also play a role in the host immunity against bacterial pathogens. Our previous study identified two type I IFNs from large yellow croaker Larimichthys croaea(Lc), LcIFNd and LcIFNh, and showed their strong induction by poly(I:C) and antiviral activities. In the present study, both LcIFNd and LcIFNh were found to be rapidly induced in head kidney and spleen by mixed bacteria of Vibrio alginolyticus, Vibrio parahaemolyticus, and Aeromonas hydrophila. In the head kidney primary cells (HKCs), expression of these two LcIFN genes was increased by peptidoglycan (PGN) from Bacillus subtilis and lipopolysaccharide (LPS) from Escherichia coli. Consistently, Lc IFN-regulatory factor (LcIRF) 3 and LcIRF7, two key transcription factors of type I IFN expression, were also induced by these three bacteria, PGN, and LPS. These observations strongly suggested that large yellow croaker type I IFNs are involved in the immune response against bacterial infection. Luciferase assays showed that promoters of both LcIFNd and LcIFNh were activated by PGN, LPS, and genomic DNA of A. hydrophila, and A. hydrophila DNA was more potent than PGN and LPS in activating LcIFNd and LcIFNh promoters. Furthermore, the induction of LcIFNd promoter by these bacterial stimuli was further enhanced by the overexpression of LcIRF7 or LcIRF7 along with LcIRF3, while that of LcIFNh promoter was increased following the overexpression of LcIRF3 alone, suggesting that the induction of these two large yellow croaker IFNs by bacterial stimuli may be regulated via distinct manners. These results therefore revealed novel aspects of the functional regulation of teleost type I IFNs.


Asunto(s)
Enfermedades de los Peces/genética , Proteínas de Peces/genética , Infecciones por Bacterias Gramnegativas/veterinaria , Interferón Tipo I/genética , Perciformes , Vibriosis/veterinaria , Aeromonas hydrophila/fisiología , Animales , ADN Bacteriano/farmacología , Enfermedades de los Peces/inmunología , Enfermedades de los Peces/microbiología , Proteínas de Peces/metabolismo , Infecciones por Bacterias Gramnegativas/genética , Infecciones por Bacterias Gramnegativas/inmunología , Infecciones por Bacterias Gramnegativas/microbiología , Riñón Cefálico/inmunología , Riñón Cefálico/metabolismo , Riñón Cefálico/microbiología , Interferón Tipo I/metabolismo , Lipopolisacáridos/farmacología , Peptidoglicano/farmacología , Regiones Promotoras Genéticas , Bazo/inmunología , Bazo/metabolismo , Bazo/microbiología , Vibriosis/genética , Vibriosis/inmunología , Vibriosis/microbiología , Vibrio alginolyticus/fisiología , Vibrio parahaemolyticus/fisiología
4.
Antimicrob Agents Chemother ; 60(1): 544-53, 2016 01.
Artículo en Inglés | MEDLINE | ID: mdl-26552982

RESUMEN

Biofilms consist of surface-adhered bacterial communities encased in an extracellular matrix composed of DNA, exopolysaccharides, and proteins. Extracellular DNA (eDNA) has a structural role in the formation of biofilms, can bind and shield biofilms from aminoglycosides, and induces antimicrobial peptide resistance mechanisms. Here, we provide evidence that eDNA is responsible for the acidification of Pseudomonas aeruginosa planktonic cultures and biofilms. Further, we show that acidic pH and acidification via eDNA constitute a signal that is perceived by P. aeruginosa to induce the expression of genes regulated by the PhoPQ and PmrAB two-component regulatory systems. Planktonic P. aeruginosa cultured in exogenous 0.2% DNA or under acidic conditions demonstrates a 2- to 8-fold increase in aminoglycoside resistance. This resistance phenotype requires the aminoarabinose modification of lipid A and the production of spermidine on the bacterial outer membrane, which likely reduce the entry of aminoglycosides. Interestingly, the additions of the basic amino acid L-arginine and sodium bicarbonate neutralize the pH and restore P. aeruginosa susceptibility to aminoglycosides, even in the presence of eDNA. These data illustrate that the accumulation of eDNA in biofilms and infection sites can acidify the local environment and that acidic pH promotes the P. aeruginosa antibiotic resistance phenotype.


Asunto(s)
Aminoglicósidos/farmacología , Antibacterianos/farmacología , Biopelículas/efectos de los fármacos , ADN Bacteriano/farmacología , Farmacorresistencia Bacteriana/efectos de los fármacos , Pseudomonas aeruginosa/efectos de los fármacos , Arabinosa/análogos & derivados , Arabinosa/metabolismo , Arginina/metabolismo , Arginina/farmacología , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Biopelículas/crecimiento & desarrollo , Transporte Biológico , ADN Bacteriano/metabolismo , Farmacorresistencia Bacteriana/genética , Espacio Extracelular/química , Regulación Bacteriana de la Expresión Génica , Concentración de Iones de Hidrógeno , Lípido A/metabolismo , Pruebas de Sensibilidad Microbiana , Plancton/efectos de los fármacos , Plancton/crecimiento & desarrollo , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/metabolismo , Bicarbonato de Sodio/metabolismo , Bicarbonato de Sodio/farmacología , Espermidina/metabolismo , Espermidina/farmacología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
5.
Genome Res ; 22(4): 802-9, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22300632

RESUMEN

In the process of clone-based genome sequencing, initial assemblies frequently contain cloning gaps that can be resolved using cloning-independent methods, but the reason for their occurrence is largely unknown. By analyzing 9,328,693 sequencing clones from 393 microbial genomes, we systematically mapped more than 15,000 genes residing in cloning gaps and experimentally showed that their expression products are toxic to the Escherichia coli host. A subset of these toxic sequences was further evaluated through a series of functional assays exploring the mechanisms of their toxicity. Among these genes, our assays revealed novel toxins and restriction enzymes, and new classes of small, non-coding toxic RNAs that reproducibly inhibit E. coli growth. Further analyses also revealed abundant, short, toxic DNA fragments that were predicted to suppress E. coli growth by interacting with the replication initiator DnaA. Our results show that cloning gaps, once considered the result of technical problems, actually serve as a rich source for the discovery of biotechnologically valuable functions, and suggest new modes of antimicrobial interventions.


Asunto(s)
ADN Bacteriano/genética , Escherichia coli/genética , Genes Bacterianos/genética , ARN Bacteriano/genética , Antibacterianos/metabolismo , Antibacterianos/farmacología , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Secuencia de Bases , Sitios de Unión/genética , Clonación Molecular , ADN Bacteriano/metabolismo , ADN Bacteriano/farmacología , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Escherichia coli/metabolismo , Regulación Bacteriana de la Expresión Génica , Genoma Bacteriano/genética , Viabilidad Microbiana/efectos de los fármacos , Viabilidad Microbiana/genética , Datos de Secuencia Molecular , Unión Proteica , ARN Bacteriano/metabolismo , ARN Bacteriano/farmacología , ARN de Transferencia/genética , ARN de Transferencia/metabolismo , ARN de Transferencia/farmacología , Homología de Secuencia de Ácido Nucleico , Transcripción Genética
6.
J Dairy Sci ; 98(1): 106-10, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25465570

RESUMEN

Some strains of Lactobacillus genus have antiproliferative activities against cancer cells. However, until now, the exact effector molecules of Lactobacillus strains with anticancer activity have not been identified. The aim of the present study was to explore which fraction of the Lactobacillus cells exerts the highest antiproliferative effect. For this purpose, the heat-killed bacterial cells, bacterial cell wall extract, and genomic DNA of 8 Lactobacillus strains were prepared to assess their antiproliferative activities against human myeloid leukemia cell lines K562. The heat-killed bacterial cells of the 8 lactobacilli strains exerted antiproliferative effect on K562 cells, and the inhibition rates exerted by the heat-killed bacterial cells of the strains G15AL, M5AL, SB31AL, SB5AL, and T3AL were significantly higher than those exerted by the cell walls and genomic DNA of the strains. The bacterial DNA of G15AL exerted higher antiproliferative effect on K562 cells. The exact effector molecules and the effect mechanism of the strains should be further explored for the application of these strains as probiotic strains or bioactive probiotic molecules.


Asunto(s)
Proliferación Celular/efectos de los fármacos , ADN Bacteriano/farmacología , Lactobacillus/química , Leucemia Eritroblástica Aguda/microbiología , Animales , Pared Celular/química , ADN Bacteriano/aislamiento & purificación , Heces/microbiología , Humanos , Células K562 , Lactobacillus/citología , Lactobacillus/genética , Leucemia Eritroblástica Aguda/patología , Leucemia Eritroblástica Aguda/prevención & control
7.
J Infect Dis ; 210(9): 1476-86, 2014 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-24812048

RESUMEN

The mechanisms underlying the immune defense by trophoblasts against pathogens remain ill defined. We demonstrated that placental cell death was increased upon in vivo exposure to Listeria monocytogenes. The death of infected cells is an important host innate defense mechanism. Meanwhile, double-stranded DNA (dsDNA) derived from intracellular bacteria or dsDNA viruses is emerging as a potent pathogen-associated molecular pattern recognized by host cells. We sought to characterize trophoblast death in response to cytosolic dsDNA challenge. Our results showed that dsDNA induced caspase-dependent and -independent cell death in human trophoblasts. However, necroptosis, a cell death pathway independent of caspase, could not be induced by dsDNA treatment, even in the presence of exogenously expressed RIPK3. L. monocytogenes-derived genomic DNA triggered a similar cell death pattern. Moreover, the cell death in response to dsDNA was IFI16 dependent. These data suggest that cytosolic dsDNA induces nonnecroptotic cell death in trophoblasts via IFI16, and this could contribute to placental barrier against infection.


Asunto(s)
Muerte Celular/efectos de los fármacos , ADN/farmacología , Listeria monocytogenes/efectos de los fármacos , Proteínas Nucleares/fisiología , Fosfoproteínas/fisiología , Trofoblastos/microbiología , Animales , Caspasas/metabolismo , Línea Celular , ADN Bacteriano/farmacología , Femenino , Humanos , Listeria monocytogenes/fisiología , Listeriosis/microbiología , Masculino , Ratones Endogámicos C57BL , Embarazo , Trofoblastos/efectos de los fármacos
8.
Antimicrob Agents Chemother ; 58(12): 7273-82, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25267673

RESUMEN

Staphylococcus epidermidis biofilm formation is responsible for the persistence of orthopedic implant infections. Previous studies have shown that exposure of S. epidermidis biofilms to sub-MICs of antibiotics induced an increased level of biofilm persistence. BODIPY FL-vancomycin (a fluorescent vancomycin conjugate) and confocal microscopy were used to show that the penetration of vancomycin through sub-MIC-vancomycin-treated S. epidermidis biofilms was impeded compared to that of control, untreated biofilms. Further experiments showed an increase in the extracellular DNA (eDNA) concentration in biofilms preexposed to sub-MIC vancomycin, suggesting a potential role for eDNA in the hindrance of vancomycin activity. Exogenously added, S. epidermidis DNA increased the planktonic vancomycin MIC and protected biofilm cells from lethal vancomycin concentrations. Finally, isothermal titration calorimetry (ITC) revealed that the binding constant of DNA and vancomycin was 100-fold higher than the previously reported binding constant of vancomycin and its intended cellular d-Ala-d-Ala peptide target. This study provides an explanation of the eDNA-based mechanism of antibiotic tolerance in sub-MIC-vancomycin-treated S. epidermidis biofilms, which might be an important factor for the persistence of biofilm infections.


Asunto(s)
Antibacterianos/farmacología , Biopelículas/efectos de los fármacos , ADN Bacteriano/farmacología , Staphylococcus epidermidis/efectos de los fármacos , Vancomicina/antagonistas & inhibidores , Antibacterianos/metabolismo , Biopelículas/crecimiento & desarrollo , Transporte Biológico/efectos de los fármacos , Compuestos de Boro/química , ADN Bacteriano/metabolismo , Colorantes Fluorescentes/química , Pruebas de Sensibilidad Microbiana , Microscopía Confocal , Solubilidad , Coloración y Etiquetado , Staphylococcus epidermidis/fisiología , Vancomicina/metabolismo , Vancomicina/farmacología
9.
Antimicrob Agents Chemother ; 58(8): 4755-61, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24913166

RESUMEN

We investigated biofilms of two pathogens, Acinetobacter baumannii and Staphylococcus aureus, to characterize mechanisms by which the extracellular polymeric substance (EPS) found in biofilms can protect bacteria against tobramycin exposure. To do so, it is critical to study EPS-antibiotic interactions in a homogeneous environment without mass transfer limitations. Consequently, we developed a method to grow biofilms, harvest EPS, and then augment planktonic cultures with isolated EPS and tobramycin. We demonstrated that planktonic cultures respond differently to being treated with different types of EPS (A. baumannii versus S. aureus) in the presence of tobramycin. By harvesting EPS from the biofilms, we found that A. baumannii EPS acts as a "universal protector" by inhibiting tobramycin activity against bacterial cells regardless of species; S. aureus EPS did not show any protective ability in cell cultures. Adding Mg(2+) or Ca(2+) reduced the protective effect of A. baumannii EPS. Finally, when we selectively digested the proteins or DNA of the EPS, we found that the protective ability did not change, suggesting that neither has a significant role in protection. To the best of our knowledge, this is the first study that demonstrates how EPS protects pathogens against antibiotics in a homogeneous system without mass transfer limitations. Our results suggest that EPS protects biofilm communities, in part, by adsorbing antibiotics near the surface. This may limit antibiotic diffusion to the bottom of the biofilms but is not likely to be the only mechanism of protection.


Asunto(s)
Acinetobacter baumannii/efectos de los fármacos , Antibacterianos/farmacología , Biopelículas/efectos de los fármacos , Staphylococcus aureus/efectos de los fármacos , Tobramicina/farmacología , Acinetobacter baumannii/química , Acinetobacter baumannii/crecimiento & desarrollo , Proteínas Bacterianas/aislamiento & purificación , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/farmacología , Biopelículas/crecimiento & desarrollo , Calcio/farmacología , Cationes Bivalentes , ADN Bacteriano/aislamiento & purificación , ADN Bacteriano/metabolismo , ADN Bacteriano/farmacología , Magnesio/farmacología , Plancton/efectos de los fármacos , Plancton/crecimiento & desarrollo , Polisacáridos Bacterianos/aislamiento & purificación , Polisacáridos Bacterianos/metabolismo , Polisacáridos Bacterianos/farmacología , Staphylococcus aureus/química , Staphylococcus aureus/crecimiento & desarrollo , Tobramicina/antagonistas & inhibidores
10.
Am J Physiol Gastrointest Liver Physiol ; 306(11): G1021-32, 2014 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-24742987

RESUMEN

Necrotizing enterocolitis (NEC) is the leading cause of death from gastrointestinal disease in premature infants and develops partly from an exaggerated intestinal epithelial immune response to indigenous microbes. There has been interest in administering probiotic bacteria to reduce NEC severity, yet concerns exist regarding infection risk. Mechanisms of probiotic activity in NEC are unknown although activation of the microbial DNA receptor Toll-like receptor-9 (TLR9) has been postulated. We now hypothesize that the Gram-positive bacterium Lactobacillus rhamnosus HN001 can attenuate NEC in small and large animal models, that its microbial DNA is sufficient for its protective effects, and that protection requires activation of the Toll-like receptor 9 (TLR9). We now show that oral administration of live or UV-inactivated Lactobacillus rhamnosus HN001 attenuates NEC severity in newborn mice and premature piglets, as manifest by reduced histology score, attenuation of mucosal cytokine response, and improved gross morphology. TLR9 was required for Lactobacillus rhamnosus-mediated protection against NEC in mice, as the selective decrease of TLR9 from the intestinal epithelium reversed its protective effects. Strikingly, DNA of Lactobacillus rhamnosus HN001 reduced the extent of proinflammatory signaling in cultured enterocytes and in samples of resected human ileum ex vivo, suggesting the therapeutic potential of this probiotic in clinical NEC. Taken together, these findings illustrate that Lactobacillus rhamnosus HN001 is an effective probiotic for NEC via activation of the innate immune receptor TLR9 and that Lactobacillus rhamnosus DNA is sufficient for its protective effects, potentially reducing concerns regarding the infectious risk of this novel therapeutic approach.


Asunto(s)
Enterocolitis Necrotizante/prevención & control , Lacticaseibacillus rhamnosus/fisiología , Probióticos/farmacología , Receptor Toll-Like 9/metabolismo , Animales , Animales Recién Nacidos , ADN Bacteriano/farmacología , Enterocolitis Necrotizante/metabolismo , Enterocolitis Necrotizante/patología , Ratones , Nacimiento Prematuro , Porcinos , Receptor Toll-Like 9/genética
11.
Antimicrob Agents Chemother ; 57(5): 2352-61, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23478967

RESUMEN

Within recent years, it has been established that extracellular DNA is a key constituent of the matrix of microbial biofilms. In addition, it has recently been demonstrated that DNA binds positively charged antimicrobials such as aminoglycosides and antimicrobial peptides. In the present study, we provide evidence that extracellular DNA shields against aminoglycosides in Pseudomonas aeruginosa biofilms. We show that exogenously supplemented DNA integrates into P. aeruginosa biofilms and increases their tolerance toward aminoglycosides. We provide evidence that biofilms formed by a DNA release-deficient P. aeruginosa quorum-sensing mutant are more susceptible to aminoglycoside treatment than wild-type biofilms but become rescued from the detrimental action of aminoglycosides upon supplementation with exogenous DNA. Furthermore, we demonstrate that exposure to lysed polymorphonuclear leukocytes, which are thought to be a source of extracellular DNA at sites of infections, increases the tolerance of P. aeruginosa biofilms toward aminoglycosides. Although biofilm-associated aminoglycoside tolerance recently has been linked to extracellular DNA-mediated activation of the pmr genes, we demonstrate that the aminoglycoside tolerance mediated by the presence of extracellular DNA is not caused by activation of the pmr genes in our P. aeruginosa biofilms but rather by a protective shield effect of the extracellular DNA.


Asunto(s)
Aminoglicósidos/metabolismo , ADN Bacteriano/metabolismo , ADN/metabolismo , Espacio Extracelular/química , Pseudomonas aeruginosa/efectos de los fármacos , Pseudomonas aeruginosa/metabolismo , Transportadoras de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/metabolismo , Aminoglicósidos/biosíntesis , Aminoglicósidos/farmacología , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Biopelículas/efectos de los fármacos , ADN/farmacología , ADN Bacteriano/farmacología , Humanos , Pruebas de Sensibilidad Microbiana , Mutación , Neutrófilos/química , Pseudomonas aeruginosa/genética , Percepción de Quorum , Transactivadores/deficiencia , Transactivadores/genética
12.
Caries Res ; 47(4): 338-45, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23486236

RESUMEN

This study assessed whether the persistence of Lactobacillus reuteri DSM 17938 and ATCC PTA 5289 in saliva could delay the regrowth of mutans streptococci (MS) after a full-mouth disinfection with chlorhexidine (CHX). A randomised, double-blind, placebo-controlled study with a 6-week intervention period and 3- and 6-month follow-up was performed. 62 healthy subjects with moderate to high counts of MS were randomly assigned to a test group (n = 32) or a placebo group (n = 30). Before onset of the intervention, subjects received two sessions of professional cleaning, flossing, and application of CHX varnish and rinsed their mouth with a CHX solution between the sessions (2 days). Thereafter, the test group used probiotic lozenges (2/day) containing L. reuteri (DSM 17938 and ATCC PTA 5289; 1 × 10(8) CFU of each strain), and the placebo group used identical lozenges lacking the lactobacilli. Saliva samples were collected and cultured onto selective media, and isolates of L. reuteri as well as DNA directly extracted from saliva were tested by polymerase chain reaction (PCR) with specific primers. Presence of salivary MS was analysed with a chair-side test. L. reuteri was frequently detected by culture during the intervention period but in only 3 test group subjects at follow-ups. Regrowth of MS statistically significantly differed depending on the presence or absence of L. reuteri DSM 17938 detected by PCR. We conclude that cultivable L. reuteri strains may only sporadically be confirmed after termination of the intervention, but subjects with PCR-detected L. reuteri demonstrated slower regrowth of MS.


Asunto(s)
ADN Bacteriano/farmacología , Limosilactobacillus reuteri/fisiología , Probióticos/farmacología , Streptococcus mutans/crecimiento & desarrollo , Adulto , Antiinfecciosos Locales/farmacología , Clorhexidina/farmacología , Recuento de Colonia Microbiana , ADN Bacteriano/análisis , Desinfección/métodos , Método Doble Ciego , Femenino , Humanos , Masculino , Antisépticos Bucales/farmacología , Saliva/microbiología , Streptococcus mutans/efectos de los fármacos , Adulto Joven
13.
Sci Rep ; 13(1): 8041, 2023 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-37198168

RESUMEN

Microorganisms do not live as dispersed single cells but rather they form aggregates with extracellular polymeric substances at interfaces. Biofilms are considered efficient life forms because they shield bacteria from biocides and collect dilute nutrients. This is a big concern in industry since the microorganisms can colonize a wide range of surfaces, accelerating material deterioration, colonizing medical devices, contaminating ultrapure drinking water, increasing energy costs and creating focus of infection. Conventional biocides that target a specific component of the bacteria are not effective in the presence of biofilms. Efficient biofilm inhibitors are based on a multitarget approach interacting with the bacteria and the biofilm matrix. Their rationale design requires a thorough understanding of inhibitory mechanisms that are still largely lacking today. Herein we uncover via molecular modelling the inhibition mechanism of cetrimonium 4-OH cinnamate (CTA-4OHcinn). Simulations show that CTA-4OH micelles can disrupt symmetric and asymmetric bilayers, representative of inner and outer bacterial membranes, following three stages: adsorption, assimilation, and defect formation. The main driving force for micellar attack is electrostatic interactions. In addition to disrupting the bilayers, the micelles work as carriers facilitating the trapping of 4OH cinnamate anions within the bilayer upper leaflet and overcoming electrostatic repulsion. The micelles also interact with extracellular DNA (e-DNA), which is one of the main components of biofilms. It is observed that CTA-4OHcinn forms spherical micelles on the DNA backbone; which hinders their ability to pack. This is demonstrated by modelling the DNA along the hbb histone-like protein, showing that in the presence of CTA-4OHcinn, DNA does not pack properly around hbb. The abilities of CTA-4OHcinn to cause cell death through membrane disruption and to disperse a mature, multi-species biofilm are also confirmed experimentally.


Asunto(s)
Desinfectantes , Micelas , Cetrimonio/farmacología , Biopelículas , Bacterias , ADN/farmacología , Desinfectantes/farmacología , ADN Bacteriano/farmacología
14.
J Leukoc Biol ; 113(5): 461-470, 2023 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-36857592

RESUMEN

Inflammation centered on non-IgE-mediated mast cell activation characterizes chronic spontaneous urticaria resistant to nonsedating H1-antihistamines. We recently uncovered a strong positive association between inflammation and the fecal Escherichia. To further explore the actions of bacterial DNA derived from Escherichia on mast cells, intestinal permeability of patients with chronic spontaneous urticaria with or without nonsedating H1-antihistamine resistance and healthy controls were determined, and LAD2 cells with knockdown of Syk, Nedd4L, or Sgk1 or with incubation of inhibitors GS9973, GSK650394, and MG132 were posttreated with btDNA. We found that (i) serum intestinal permeability indices and bacterial DNA markedly increased in patients with chronic spontaneous urticaria with nonsedating H1-antihistamine resistance compared with those without (all P < 0.001), and bacterial DNA positively correlated with the degree of inflammation; (ii) IL-6 and TNF-α levels were time- and dose-dependently upregulated in bacterial DNA-stimulated LAD2 cells, which relied on unmethylated CpG in bacterial DNA and Toll-like receptor 9 protein in cells; (iii) Syk knockdown or inhibition of Syk Tyr525/526 phosphorylation blocked bacterial DNA-initiated cytokine production; (iv) Nedd4L interacted with Tyr525/526-phosphorylated Syk, and inhibition of Nedd4L Ser448 phosphorylation induced by bacterial DNA-activated Sgk1 was mandatory for bacterial DNA's proinflammatory property; and (v) Sgk1 suppression showed an inhibitory effect on bacterial DNA-induced inflammation by ensuring Nedd4L-mediated ubiquitination of Tyr525/526-phosphorylated Syk. Collectively, we identified previously unknown contributory roles of bacterial translocation and serum bacterial DNA on the inflammation phenotype in patients with chronic spontaneous urticaria with nonsedating H1-antihistamine resistance and further uncovered a vital negative regulatory role for the Sgk1/Nedd4L/Syk pathway in bacterial DNA-induced inflammation in LAD2 cells.


Asunto(s)
Urticaria Crónica , ADN Bacteriano , Mastocitos , Urticaria , Humanos , ADN Bacteriano/farmacología , Antagonistas de los Receptores Histamínicos , Inflamación/microbiología , Mastocitos/metabolismo , Quinasa Syk , Urticaria/tratamiento farmacológico , Urticaria/metabolismo , Urticaria/microbiología
15.
J Immunol ; 185(12): 7367-73, 2010 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-21057083

RESUMEN

Bacterial DNA contains unmethylated CpG dinucleotides and is a potent ligand for TLR9. Bacterial DNA has been claimed the active ingredient in bacterial lysates used for immunotherapy. Whereas the detection of viral DNA by TLR9 expressed in plasmacytoid dendritic cells (PDCs) with subsequent IFN-α production is well defined, the role of bacterial DNA during microbial infection is less clear. In fact, IFN-α is not a hallmark of antibacterial immune responses. Unlike in mice, TLR9 expression in humans is restricted to PDCs and B cells; thus, conclusions from murine models of infection have limitations. In this study, we demonstrate that lysates of heat-killed Escherichia coli containing bacterial DNA induced IFN-α in isolated PDCs but not in the mixed cell populations of human PBMCs. Depletion of monocytes restored IFN-α secretion by PDCs within PBMCs. We found that monocyte-derived IL-10 and PGs contribute to monocyte-mediated inhibition of IFN-α release in PDCs. We conclude that human PDCs can be stimulated by bacterial DNA via TLR9; however, in the physiological context of mixed-cell populations, PDC activation is blocked by factors released from monocytes stimulated in parallel by other components of bacterial lysates such as LPS. This functional repression of PDCs by concomitantly stimulated monocytes avoids production of antiviral IFN-α during bacterial infection and thus explains how the innate immune system is enabled to distinguish bacterial from viral CpG DNA and thus to elicit the appropriate responses despite the presence of CpG DNA in both types of infection.


Asunto(s)
ADN Bacteriano/inmunología , Células Dendríticas/inmunología , Escherichia coli K12/inmunología , Interferón-alfa/inmunología , Monocitos/inmunología , Células Plasmáticas/inmunología , Receptor Toll-Like 9/inmunología , Animales , ADN Bacteriano/química , ADN Bacteriano/farmacología , ADN Viral/química , ADN Viral/inmunología , ADN Viral/farmacología , Células Dendríticas/metabolismo , Escherichia coli K12/química , Humanos , Interferón-alfa/biosíntesis , Lipopolisacáridos/química , Lipopolisacáridos/inmunología , Lipopolisacáridos/farmacología , Ratones , Monocitos/metabolismo , Células Plasmáticas/metabolismo , Receptor Toll-Like 9/metabolismo , Virus/química , Virus/inmunología
16.
J Immunol ; 185(2): 1186-95, 2010 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-20566831

RESUMEN

Listeria monocytogenes invades the cytoplasm of macrophages and induces the activation of caspase-1 and the subsequent maturation of IL-1beta and IL-18. Although apoptosis-associated speck-like protein containing a caspase-activating and recruitment domain (ASC), an adaptor protein of nucleotide-binding oligomerization domain (Nod)-like receptors, has been shown to play an essential role in inducing this cellular response to L. monocytogenes, the mechanism has not been fully elucidated. In this study, we demonstrate the role of absent in melanoma 2 (AIM2), a recently described receptor of cytosolic DNA, in the activation of caspase-1 upon infection with L. monocytogenes. Secretion of IL-1beta and IL-18 from Nod-like receptor family, pyrin domain containing 3 (NLRP3) and Nod-like receptor family, caspase-activating and recruitment domain containing 4 (NLRC4) knockout macrophages in response to L. monocytogenes was only slightly decreased compared with the levels secreted from wild-type macrophages, whereas secretion from ASC knockout macrophages was completely impaired, suggesting that receptors other than NLRP3 and NLRC4 also take part in inflammasome activation in an ASC-dependent manner. To identify such receptors, the abilities of several receptor candidates (NLRP2, NLRP6, NLRP12, and AIM2) to induce the secretion of IL-1beta in response to L. monocytogenes were compared using the inflammasome system reconstructed in HEK293 cells. Among these receptor candidates, AIM2 conferred the highest responsiveness to the bacterium on HEK293 cells. Knockdown of AIM2 significantly decreased the secretion of IL-1beta and IL-18 from L. monocytogenes-infected macrophages. These results suggest that AIM2, in cooperation with NLRP3 and NLRC4, plays an important role in the activation of caspase-1 during L. monocytogenes infection.


Asunto(s)
Listeria monocytogenes/fisiología , Macrófagos/microbiología , Proteínas Nucleares/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Western Blotting , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Caspasa 1/genética , Caspasa 1/metabolismo , Línea Celular , Células Cultivadas , ADN Bacteriano/genética , ADN Bacteriano/inmunología , ADN Bacteriano/farmacología , Proteínas de Unión al ADN , Femenino , Interacciones Huésped-Patógeno/efectos de los fármacos , Interacciones Huésped-Patógeno/inmunología , Humanos , Interleucina-18/genética , Interleucina-18/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Listeria monocytogenes/genética , Listeria monocytogenes/inmunología , Macrófagos/citología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR , Proteínas Nucleares/genética , Fagosomas/inmunología , Fagosomas/metabolismo , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
17.
Rheumatology (Oxford) ; 50(4): 689-96, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21149241

RESUMEN

OBJECTIVES: CpG motifs, which are highly prevalent in bacterial DNA, have been shown to trigger the production of ANCA in vitro by B lymphocytes from patients with active ANCA-associated vasculitis (AAV). Staphylococcus aureus is associated with relapses in AAV, and CpG motifs from staphylococcal DNA may trigger ANCA production in AAV patients in remission. We investigated the presence of ANCA-producing B lymphocytes during quiescent disease and tested the capacity of these cells to produce ANCA in response to CpG. METHODS: Expression of Toll-like receptor 9 (TLR9) by B lymphocytes from AAV patients and controls was assessed. Peripheral blood mononuclear cells were isolated from 23 PR3-ANCA and 15 MPO-ANCA patients (33 quiescent, 5 active disease) and 14 healthy controls, and cultured for 12 days in the presence of cytosine-phosphate-guanine oligodeoxynucleotide (CpG-ODN) and IL-2. B-lymphocyte activation, differentiation, immunoglobulin production and in vitro ANCA production were studied. RESULTS: TLR9 expression by B lymphocytes was comparable in AAV patients and controls. B lymphocytes were activated and differentiated towards a plasma cell phenotype in response to CpG-ODN and IL-2. ANCA were produced in vitro by 13 out of 23 PR3-ANCA patients and 3 out of 15 MPO-ANCA patients. CONCLUSIONS: We conclude that ANCA-producing B lymphocytes can be present in the peripheral blood of AAV patients during remission. These autoreactive B lymphocytes are triggered by CpG-ODN and IL-2 to produce ANCA in vitro. CpG motifs may trigger the production of ANCA in vivo, contributing to the development of relapses in AAV.


Asunto(s)
Vasculitis Asociada a Anticuerpos Citoplasmáticos Antineutrófilos/metabolismo , Anticuerpos Anticitoplasma de Neutrófilos/metabolismo , Linfocitos B/metabolismo , ADN Bacteriano/farmacología , Oligodesoxirribonucleótidos/farmacología , Remisión Espontánea , Adulto , Anciano , Vasculitis Asociada a Anticuerpos Citoplasmáticos Antineutrófilos/etiología , Vasculitis Asociada a Anticuerpos Citoplasmáticos Antineutrófilos/patología , Linfocitos B/efectos de los fármacos , Linfocitos B/patología , Estudios de Casos y Controles , Células Cultivadas , Femenino , Humanos , Interleucina-2/farmacología , Recuento de Linfocitos , Masculino , Persona de Mediana Edad , Fenotipo , Recurrencia , Staphylococcus aureus/genética , Receptor Toll-Like 9/metabolismo
18.
J Clin Immunol ; 30(5): 693-702, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20490635

RESUMEN

Infection with Listeria monocytogenes can cause meningitis and septicemia in newborn, elderly, or immunocompromised individuals. Pregnant women are particularly susceptible to Listeria, leading to a potentially fatal infection. Cytosolic Listeria activates the proinflammatory caspase-1 and induces the processing and secretion of interleukins IL-1beta and IL-18 as well as caspase-1-dependent pyroptosis. This study elucidates the role of various inflammasome components of host macrophages in the proinflammatory response to infection with Listeria. Here, we have used macrophages from AIM2-, NLRC4-, NLRP3-, and ASC-deficient mice to demonstrate that AIM2, NLRC4, and NLRP3 inflammasomes as well as the adaptor protein ASC all contribute to activation of caspase-1 in Listeria-infected macrophages. We show that Listeria DNA, which escapes into the cytosol of infected macrophages, triggers AIM2 oligomerization, caspase-1 activation, and pyroptosis. Interestingly, we found that flagellin-deficient Listeria, like Francisella tularensis, is recognized primarily by the AIM2 inflammasome, as no caspase-1 activation or cell death was observed in AIM2-deficient macrophages infected with this Listeria mutant. We further show that prior priming of NLRC4-deficient macrophages with LPS is sufficient for Listeria-induced caspase-1 activation in these macrophages, suggesting that TLR4 signaling is important for activation of the AIM2 and NLRP3 inflammasomes by Listeria in the absence of NLRC4. Taken together, our results indicate that Listeria infection is sensed by multiple inflammasomes that collectively orchestrate a robust caspase-1 activation and proinflammatory response.


Asunto(s)
Inflamasomas/inmunología , Interleucina-1beta/metabolismo , Listeria monocytogenes/inmunología , Listeriosis/inmunología , Macrófagos/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Caspasa 1/metabolismo , Línea Celular Transformada , ADN Bacteriano/farmacología , Proteínas de Unión al ADN , Activación Enzimática/efectos de los fármacos , Activación Enzimática/genética , Flagelina/genética , Flagelina/inmunología , Interleucina-1beta/inmunología , Listeria monocytogenes/genética , Listeria monocytogenes/patogenicidad , Listeriosis/genética , Listeriosis/metabolismo , Macrófagos/inmunología , Macrófagos/patología , Ratones , Ratones Noqueados , Mutación/genética , Proteína con Dominio Pirina 3 de la Familia NLR , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Multimerización de Proteína/efectos de los fármacos , Multimerización de Proteína/genética
19.
PLoS Pathog ; 4(11): e1000213, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19023416

RESUMEN

Biofilms are surface-adhered bacterial communities encased in an extracellular matrix composed of DNA, bacterial polysaccharides and proteins, which are up to 1000-fold more antibiotic resistant than planktonic cultures. To date, extracellular DNA has been shown to function as a structural support to maintain Pseudomonas aeruginosa biofilm architecture. Here we show that DNA is a multifaceted component of P. aeruginosa biofilms. At physiologically relevant concentrations, extracellular DNA has antimicrobial activity, causing cell lysis by chelating cations that stabilize lipopolysaccharide (LPS) and the outer membrane (OM). DNA-mediated killing occurred within minutes, as a result of perturbation of both the outer and inner membrane (IM) and the release of cytoplasmic contents, including genomic DNA. Sub-inhibitory concentrations of DNA created a cation-limited environment that resulted in induction of the PhoPQ- and PmrAB-regulated cationic antimicrobial peptide resistance operon PA3552-PA3559 in P. aeruginosa. Furthermore, DNA-induced expression of this operon resulted in up to 2560-fold increased resistance to cationic antimicrobial peptides and 640-fold increased resistance to aminoglycosides, but had no effect on beta-lactam and fluoroquinolone resistance. Thus, the presence of extracellular DNA in the biofilm matrix contributes to cation gradients, genomic DNA release and inducible antibiotic resistance. DNA-rich environments, including biofilms and other infection sites like the CF lung, are likely the in vivo environments where extracellular pathogens such as P. aeruginosa encounter cation limitation.


Asunto(s)
Biopelículas , Cationes/farmacología , ADN Bacteriano/farmacología , Farmacorresistencia Microbiana , Pseudomonas aeruginosa/fisiología , Péptidos Catiónicos Antimicrobianos , Proteínas Bacterianas , ADN Bacteriano/ultraestructura , Lipopolisacáridos , Factores de Transcripción
20.
Appl Environ Microbiol ; 76(9): 2846-55, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20208019

RESUMEN

The beneficial effects of Bifidobacterium are partly due to its immunostimulatory properties. These immunostimulatory properties may be linked to the presence of unmethylated CpG motifs specific to bacterial DNA, which may induce a TH1 response by activating Toll-like receptors (TLR). Using in silico analyses, PCR amplification, and dot blotting, we characterized the CpG content of various bifidobacterial strains and evaluated the immunostimulatory properties and genomic heterogeneity of these motifs in the genus. Our in silico study, based on entire genome sequences from five bifidobacterial strains, showed that Bifidobacterium genomes contain numerous CpG motifs, including 5'-purine-purine-CG-pyrimidine-pyrimidine-3' and 5'-purine-TCG-pyrimidine-pyrimidine-3' motifs, and biologically active sequences previously identified in lactic acid bacteria. We identified four CpG-rich sequences with Bifidobacterium longum NCC2705. Two sequences with a percent G+C of about 68% included 14 and 16 CpG motifs. Two sequences with a percent G+C of about 60% included 16 and 6 CpG motifs. These sequences induce the production of monocyte chemoattractant protein 1 (MCP-1) and tumor necrosis factor alpha (TNF-alpha) through a pattern of TLR9 stimulation on RAW 264.7 macrophages. No link could be established between their immunostimulatory properties, the number of CpG motifs, and percent G+C. We investigated inter- and intraspecies heterogeneity in 71 strains of various origins. These sequences were highly conserved in the genus. No link was found between the presence of the CpG-rich sequence and the origin of the strains (healthy, allergic, or preterm infants). The high frequency of CpG motifs in the DNA of Bifidobacterium may play an important role in the immunostimulatory properties of commensal or probiotic bifidobacterial strains.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Bifidobacterium/genética , Islas de CpG , ADN Bacteriano/farmacología , Fosfatos de Dinucleósidos/farmacología , Adyuvantes Inmunológicos/genética , Secuencia de Bases , ADN Bacteriano/química , Humanos , Lactante , Receptores Toll-Like/metabolismo , Factor de Necrosis Tumoral alfa/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA